You are on page 1of 13

REVIEW

How Do MicroRNAs Regulate


Gene Expression?
Richard J. Jackson* and Nancy Standart*
(Published 2 January 2007)

Several thousand human genes, amounting to about and reviews written before that date generally proposed that
one-third of the whole genome, are potential targets miRNAs (i) do not promote degradation of their target mRNAs,
for regulation by the several hundred microRNAs but (ii) they do down-regulate target mRNA translation at some
(miRNAs) encoded in the genome. The regulation stage after the initiation step.
occurs posttranscriptionally and involves the ~21- Over the past 18 months, there has been a steady flow of
nucleotide miRNA interacting with a target site in the provocative reports on mechanisms of miRNA-mediated repres-
mRNA that generally has imperfect complementarity sion, and, far from consolidating and building upon these earlier
to the miRNA. The target sites are almost invariably ideas, many of them directly contradict one or another of these
in the 3′-untranslated region of the messenger RNA original assertions. The aim of this Review is to illuminate and
(mRNA), often in multiple copies. Metazoan miRNAs explain the controversies generated by these recent publications.
were previously thought to down-regulate protein However, we have been unable to resolve many of the apparent

Downloaded from stke.sciencemag.org on February 26, 2009


expression by inhibiting target mRNA translation at contradictions, and our advice to the reader is to keep an open
some stage after the translation initiation step, with- mind to the possibility that there may be more than one mecha-
out much effect on mRNA abundance. However, nism by which miRNAs effect posttranscriptional regulation of
recent studies have questioned these suppositions. gene expression.
With some targets, an increase in the rate of mRNA This Review will therefore focus specifically on mechanisms,
degradation by the normal decay pathway con- with particular reference to metazoans. Lower eukaryotes are
tributes to the decrease in protein expression. mentioned only to the extent that they have contributed to our
miRNAs can also inhibit translation initiation, specif- understanding of mechanisms likely to operate in higher organ-
ically the function of the cap-binding initiation factor, isms, and plants are ignored altogether, as the mechanisms of
eIF4E. Repressed target mRNAs as well as miRNAs plant miRNA biogenesis and action seem distinctly different.
themselves accumulate in cytoplasmic foci known Mechanisms of RNA interference (RNAi) by short interfering
as P-bodies, where many enzymes involved in mRNA RNAs (siRNAs) are also beyond the scope of this Review except
degradation are concentrated. However, P-bodies insofar as they relate directly to miRNA-mediated repression.
may also serve as repositories for the temporary and
reversible storage of untranslated mRNA, and reduc- siRNAs Versus miRNAs: Definitions, Similarities, and
ing the expression (knockdown) of several distinct Differences
P-body protein components can alleviate miRNA- The generally accepted distinction between “miRNAs” and
mediated repression of gene expression. “siRNAs” is that the former are ~21-residue RNAs derived
from longer RNAs that include a ~70-nucleotide (nt) imperfect-
Introduction ly base-paired hairpin segment that is the precursor of the ma-
A PubMed search of “microRNA” or “miRNA” gives just over ture miRNA, whereas siRNAs are of similar length but are de-
1200 citations, all of them published since the turn of the mil- rived from longer, perfectly complementary double-stranded
lennium. These numerous publications have been concerned RNA precursors of endogenous or exogenous origin (5). In both
primarily with using experimental and in silico approaches to cases, biogenesis proceeds through a staggered duplex interme-
identify miRNAs and the target genes that they regulate in vari- diate, usually with ~19 base pairs (bp), and always with un-
ous organisms, elucidating the biogenesis of miRNAs, and paired 3′ extensions of 2 nt and a 5′-phosphate on each strand.
investigating how the spectrum of miRNAs differs in different Especially in the case of mammalian cells, these ~21-nt RNAs
tissues and disease states. Thus, by mid-2005, it was known that are often introduced by transfection of chemically synthesized
the human genome encodes several hundred different versions of this staggered duplex RNA, or alternatively as DNA
microRNAs (1) and that the pattern of miRNA expression is constructs that will give rise to such a duplex after transcription
often perturbed in disease states (2–4). Bioinformatic approaches and processing. One of the two strands, the one with the lower-
(1) further suggested that the mammalian miRNA repertoire stability base-pairing at its 5′ end, is then assimilated into a
might collectively regulate several thousand genes, even though complex with several proteins, while the other, the so-called
only a handful of these predicted targets have been validated to passenger strand, is degraded [reviewed in (6)]. The resulting
date. By contrast, very few of these studies addressed the mech- RNA-protein complex is generally known as RISC (RNA-
anism of miRNA-mediated repression. Primary publications induced silencing complex), or for miRNAs, it is sometimes
known as an miRNP (miRNA-protein complex).
Department of Biochemistry, University of Cambridge, 80 Tennis For RNAi, the 21-residue siRNA, or its longer double-stranded
Court Road, Cambridge CB2 1GA, UK. RNA precursor, is designed to be perfectly complementary to
*E-mail: rjj@mole.bio.cam.ac.uk (R.J.J.); nms@mole.bio.cam.ac.uk part of the target mRNA sequence, which leads to endonucle-
(N.S.) olytic cleavage of the mRNA at the site of complementarity (6),

www.stke.org/cgi/content/full/2007/367/re1 Page 1
REVIEW

target sites are almost invariably located in the 3′-untranslated


A
region (3′-UTR) of a gene, often in multiple copies.
A A It is important to note that the formal distinction between an
mRNA 5’ ...AAGUUUUCAC AGCUAACA... 3’
CXCR4 3’ UUCAAAAGUG UCGAUUGUp 5’
siRNA and an miRNA is based solely on their different origins
A G and biogenesis, irrespective of the degree of complementarity to
G the target site and irrespective of whether the ~21-nt RNA pro-
G motes endonucleolytic cleavage of the target mRNA, or repres-
U A
U A sion of the target by some other mechanism. According to these
RL mRNA 5’ ...GCACAGCCUA CUACCUCA... 3’ definitions, a ~21-nt RNA introduced into cells by transfecting
let-7a 3’ UAUGUUGGAU GAUGGAGUp 5’ a chemically synthesized staggered duplex intermediate should
UUGG
be called an “siRNA,” even if its sequence has been designed to
B be precisely the same as that of a natural miRNA. This is a po-
tential source of confusion to those outside the immediate field,
...G U G U UA... and in order to avoid such difficulties, it might be preferable to
lin-28 5’ CAC GU UCUCAGG 3’ refer to such an RNA as a si(mi)RNA, to highlight the fact that
miR-125b 3’ GUG CA AGAGUCC 5’
A U U A C
C CUp although it is technically an siRNA, in fact it is one that is de-
UC signed to exactly mimic a bona fide miRNA.
Because the different outcomes depend on the degree of com-
plementarity between the short RNA and its target, it follows that
(C. e.) lin-14 5’ ...UC UAC CUCAGGGA C... 3’ a ~21-nt RNA designed to promote endonucleolytic cleavage of a

Downloaded from stke.sciencemag.org on February 26, 2009


3’ AG GUG GAGUCCCU Up 5’
lin-4 U A C
A particular mRNA with a perfectly complementary target site
A C could also act as a translational repressor of another mRNA if
CU
this has appropriate mismatched bulged target sites. The validity
U of this argument has been demonstrated for an siRNA designed
A U
5’ ...UUAUACAAC CUGCCUC... 3’ for RNAi of mRNA encoding the CXCR4 chemokine receptor
(C. e.) lin-41
let-7 3’ GAUAUGUUG GAUGGAG 5’ (8). This siRNA has subsequently been widely used to study how
U A U Up interaction of a ~21-nt RNA with imperfectly complementary 3′-
UTR sites results in repression of mRNA translation. For the rea-
Fig. 1. Examples of the imperfect complementarity between vari- sons explained above and because the sequence of this small
ous miRNAs and their bulged target sites. Micro-RNA sequences RNA shows no resemblance to any known natural miRNA, we
are in red, mRNA target sites in green. The two miRNA/mRNA will refer to it as CXCR4 siRNA, but, except where otherwise
pairs in (A) have been widely used in studies of the mechanisms stated, it should be understood that it is always being used in con-
of miRNA-mediated repression discussed here. The first is an junction with mismatched target sites and is therefore serving as
artificial system based on an siRNA designed for RNAi of
a surrogate for a typical microRNA.
CXCR4 mRNA, but adapted to serve as a miRNA mimic through
The mismatch between an miRNA and its target site can take
the design of bulged target sites in the reporter mRNA 3′-UTR.
The second is the interaction of (endogenous) let-7a miRNA with
on different configurations, with a central unpaired bulge in ei-
a reporter target site based on the predicted interaction of let-7a ther the miRNA or mRNA strand, or both. Figure 1 shows both
with human lin-28 mRNA. The three examples in (B) are natural miRNAs paired to their natural target, and the pairing of
endogenous miRNAs and their predicted (miR-125b/human the artificial CXCR4 siRNA with its laboratory-designed target
lin-28 mRNA) or validated (C. elegans lin-4 and let-7 miRNAs) site. In the latter case, the exact configuration of the unpaired
targets sites. Relevant references cited in this work are as bulge(s) affects the degree of repression, but this may not be ap-
follows (note that these are not necessarily the publications that plicable for other miRNA-mRNA pairs (9). Two extensive stud-
first identified the miRNA/target site interaction): mRNA/CXCR4 ies of how target site sequence and complementarity influence
(8, 11, 12, 33, 79); RL mRNA/let-7a (29); lin-28/miR-125b (47); repression by either natural Drosophila miRNAs overexpressed
(C.e.) lin-14/lin-4 (22–25, 49); (C.e.) lin-41/let-7 (49). in the wing imaginal disc or by the CXCR4 siRNA transfected
into HeLa cells are in very close agreement as to what is impor-
followed by degradation of the two fragments by enzymes of the tant (10, 11). First and foremost, residues in the 5′ portion of the
normal mRNA decay pathways. Cleavage may not be entirely miRNA (residues 2 to 8, the so-called seed) upstream of the dis-
abrogated by a very limited number of mismatches, depending continuity should be (almost) perfectly complementary to the
on their position, and so miRNAs that have a similar near-per- mRNA. The degree of repression is dependent on the stability of
fect complementarity to their target sites can also promote the pairing in this region, with a rather sharp cut-off, and in ad-
degradation by this route (7). Generally, however, the interaction dition, A-form geometry seems important because G-U pairs in
of an miRNA with its predicted or validated target involves quite this region decrease repression to a greater extent than can be
extensive mismatched bulges, especially in the central region explained purely on the grounds of mRNA stability. If the 5′ por-
(Fig. 1), and to a lesser extent in the miRNA 3′ end. This type of tion of the miRNA is optimally base-paired, pairing between the
interaction does not generally lead to mRNA degradation via en- 3′ portion and the mRNA target is not critical, but a high degree
donucleolytic cleavage, but results in a decrease in protein ex- of complementarity here can rescue repression when the pairing
pression that is usually (but not always) greater than the decrease of the miRNA “seed” is marginally suboptimal. In the CXCR4
in mRNA abundance, suggesting that the main reason for down- system, the degree of repression is related to the number of tar-
regulation is reduced efficiency of translation rather than in- get sites, at least up to six such sites, the maximum tested so far
creased mRNA degradation. Validated and predicted miRNA (8, 12). In addition, different miRNAs can act in a combinatorial

www.stke.org/cgi/content/full/2007/367/re1 Page 2
REVIEW

way, in the sense that cotransfection of two different siRNAs the L1 larval stage, and by the late L2 or early L3 stage, the
with a reporter construct that had two 3′-UTR bulged target sites amounts of LIN-14 and LIN-28 proteins are less than 10% of
for each of them resulted in a similar degree of repression, as that seen in mid-L1, but the abundance of lin-28 mRNA is es-
when there were four identical target sites for just one of the siRNAs sentially unchanged, whereas that of lin-14 mRNA is about half
(11). Many endogenous mRNAs have just one or two predicted of that in the L1 stage (24, 25), with no appreciable difference
sites for interaction with endogenous miRNAs, yet seem to be in poly(A) tail length (15 to 30 A residues).
quite efficiently repressed, which raises questions as to whether In sucrose gradient analyses of polysomes at the late L2 or ear-
the artificial systems such as CXCR4 may be intrinsically less ly L3 stage, the repressed lin-14 and lin-28 mRNAs were found in
potent repressors in some way. This idea is given some support the same fractions as in a polysome distribution analysis of
by recent indications that the sequences flanking miRNA target mid-L1 larvae, where both would be efficiently translated (24,
sites, or the context of such sites, can influence the biological 25). When these polysomes from late L2 larvae were analyzed on
outcome of miRNA-mRNA interactions (13). gradients containing EDTA, both mRNAs were found near the top
The contrasting outcomes that are dependent on whether of the gradient, and on metrizamide gradients the lin-14 mRNA
interaction of the ~21-nt small RNA with the mRNA target site was found in fractions of the same buoyant density as those con-
is perfectly complementary or has substantial bulged mismatches taining polysomes from L1 larvae (24, 25). Thus, by these two
are presumably due to different proteins being present in the commonly used criteria, lin-14 and lin-28 mRNAs appeared to be
RISC and miRNP or, if the protein composition is really identi- genuinely in polysomes in late L2 and early L3 larvae, even
cal, then to the degree of complementarity influencing how though their translation was repressed at these stages. These tests
these proteins act. The protein compositions of RISC assembled do not, however, prove that the polysomes are dynamic and capa-
on perfectly complementary siRNAs of exogenous origin, and ble of elongation, an issue that can be examined by seeing whether

Downloaded from stke.sciencemag.org on February 26, 2009


the miRNPs formed with endogenous miRNAs, are often the repressed mRNA moves into smaller polysomes on incubation
described as similar or overlapping rather than absolutely iden- with puromycin, or with a specif ic inhibitor of initiation
tical. However, as the functional distinction between between a (polysome runoff assays). These functional tests are best done in
perfectly complementary interaction and one with mismatches intact cells, in case the translational repression might be relieved
is only apparent after the ~21-nt RNA has interacted with the by the mere act of preparing a cell-free extract, and should be
target site, any differences are likely to arise concomitantly done over a relatively short time period (~5 min) so that differences
with, or shortly after, the establishment of this interaction. in the rate of reaction with puromycin or the rate of runoff can be
Both types of complex include members of the Argonaute detected. However, puromycin and specific initiation inhibitors do
family of proteins. Of the four mammalian Argonaute proteins, not penetrate C. elegans larvae, and so the best that could be done
only Ago2 is capable of siRNA-mediated endonucleolytic cleav- was to isolate polysomes from early L3 larvae and add them to
age, with its ribonuclease H (RNase H)–like domain cutting the reticulocyte lysate for polysome runoff, which showed that in a
phosphodiester bond in the target mRNA opposite the 10th and rather long incubation period (45 min), the lin-28 mRNA moved
11th residue in the siRNA (14, 15). Cocrystal structures show out of polysomes as effectively as did an mRNA that was not
that the 5′-phosphate on the siRNA is essential to place this repressed in the L3 larvae (25). The question of whether this
phosphodiester bond in the active site (16, 17). Such endonucle- runoff produced any full-length LIN-28 protein was not examined.
olytic cleavage would not be expected with most miRNA-mRNA The results described above are not a peculiarity of C. elegans
interactions because the A-form geometry would be severely dis- larval development. Similar results were obtained when DNA
rupted by the unpaired bulges in this region (Fig. 1). transfections of 293T cells were used to study the repression of a
Knockdown (via RNAi) of each Ago protein individually in luciferase reporter with six bulged sites for interaction with
human embryonic kidney (HEK) 293 cells showed that Ago2 CXCR4 siRNA. Introduction of the CXCR4 siRNA reduced
was the major contributor to miRNA-mediated repression of overall luciferase expression by more than 95%, with relatively
target mRNA translation (18). Nevertheless, there is evidence little effect on target mRNA abundance (12). Likewise, the pres-
that Ago1, Ago3, and Ago4 are each capable of promoting this ence or absence of the CXCR4 siRNA had no influence on the
repression to some extent (14, 19), even though they have no polysomal distribution of the target mRNA, nor did it affect the
known enzymatic activity. In contrast, the two Argonaute movement of this mRNA into smaller polysomes on a brief
proteins in Drosophila appear to have largely nonoverlapping (3 min) incubation of the cells with puromycin. On a 5-min incu-
functions, with Ago1 involved mainly or exclusively in miRNA- bation with a specific inhibitor of initiation, the target mRNA
mediated repression, and Ago2 restricted to siRNA-mediated moved into smaller polysomes, as expected, but the shift was dis-
endonucleolytic cleavage (20, 21). tinctly greater under conditions of repression by CXCR4 siRNA.
This was interpreted as premature ribosome drop-off, or a failure
Evidence That miRNAs Can Inhibit Translation at of processivity (12). However, if such drop-off was occurring
Some Stage After Initiation continuously, it is hard to see why the steady-state polysomal dis-
The pre-2005 hypothesis that miRNA-mediated repression of tribution of the target mRNA under repressed conditions would
target gene expression is due to inhibition of mRNA translation be so similar to the unrepressed distribution, unless there was a
at some stage after the initiation step was based on just two fortuitous increase in initiation frequency that precisely compen-
studies of a single Caenorhabditis elegans miRNA, lin-4 miRNA sated for the decrease in ribosome transit time. A plausible alter-
(the first microRNA to be discovered, long before the designa- native explanation is that the target mRNA is slightly more sensitive
tion “microRNA” had been invented), and two larval mRNA to the initiation inhibitor when it is repressed by the CXCR4
targets, lin-14 mRNA, which has up to seven potential 3′-UTR siRNA than it is under unrepressed conditions.
sites for bulged interaction with lin-4 miRNA (22, 23), and In this system, both cistrons of a dicistronic reporter with
lin-28 mRNA. Expression of lin-4 miRNA commences late in either the hepatitis C virus (HCV) or cricket paralysis virus

www.stke.org/cgi/content/full/2007/367/re1 Page 3
REVIEW

(CrPV) IRES (internal ribosome entry site) driving downstream authors found a polysome distribution of the repressed mRNA
cistron translation were susceptible to repression by CXCR4 suggestive of inhibited initiation (see below), and thus it remains
siRNA, with the downstream cistron possibly even more sensi- a possibility that cotranslational degradation of the nascent pro-
tive than the upstream one (12). Because initiation driven by the tein might occur specifically in those experimental conditions
CrPV IRES occurs by a highly unusual mechanism not requir- where the repressed mRNA cosediments with large polysomes.
ing any of the canonical translation initiation factors (26), this
result is consistent with the view that the repression mechanism Evidence That miRNAs Can Inhibit Initiation Depen-
does not operate at the stage of initiation, but at some later step. dent on eIF4E
Cosedimentation of miRNAs with polysomes has been taken The idea that miRNAs regulate mRNA translation at some stage
as further evidence that repression occurs on target mRNAs after the initiation step was challenged by the publication of two
when they are actually in polysomes, and this is a valid papers about a year ago that point toward initiation as the regu-
argument in cases where most of the cellular complement of lated step. The first, by Pillai et al., studied repression by
a particular miRNA is polysome-associated. However, if there endogenous let-7a miRNA in HeLa cells, using luciferase
is only a minor fraction cosedimenting with polysomes, this reporters with three bulged target sites in the 3′-UTR, with
could possibly represent miRNAs associated with those mRNA sequestration of let-7a using an antisense 2′-O-methyl oligonu-
molecules that are not fully repressed. In late L2 stage C. elegans cleotide serving as a control (29). In DNA transfections,
larvae, some 10% of the total lin-4 miRNA cosedimented with luciferase expression was reduced by 80 to 90% at 48 hours
polysomes (24). Similarly, a small fraction of the miR-124a after transfection, but reporter mRNA abundance was reduced
present in a human neuronal cell line cosedimented with by 20%. Polysome prof iles showed a distribution of the
polysomes in an EDTA-sensitive manner (27), and a more repressed mRNA that was skewed toward the top of the gradi-

Downloaded from stke.sciencemag.org on February 26, 2009


substantial fraction of this and seven other miRNAs examined ent, peaking in the 40S to disome region, strongly indicative of
was polysome-associated in rat forebrain neurons (28). inhibited initiation (29). A similar sucrose gradient distribution
It is difficult to produce an explanation for an mRNA having of endogenous cationic amino acid transporter–1 mRNA was
almost exactly the same polysomal distribution under condi- seen under conditions where it was repressed by endogenous
tions of both strong miRNA-mediated repression and no repres- miR-122 in Huh7 cells, which shows that this distribution is not
sion. As Olsen and Ambros pointed out when this was first a peculiarity of overexpressed luciferase reporter targets (30).
discovered for lin-14 mRNA, inhibition of termination or an In direct RNA transfections assayed at 6 hours after transfec-
inhibition of elongation that is less than 100% complete would tion, 7-methylguanosine (m7GpppG)–capped polyadenylated
cause polysomes to increase in size (24). Conversely, an inhibi- mRNAs (where ppp represents a triphosphate moiety) that
tion of elongation that is literally 100% complete will freeze would be translated by the scanning mechanism were suscepti-
preexisting mRNA in polysomes, but any newly synthesized ble to repression by let-7a, and were only slightly less sensitive
mRNA will become associated with only a monomeric 80S to this repression if the poly(A) tail was absent. By contrast, a
ribosome. In the context of miRNA regulation of translation, m7GpppG-capped polyadenylated reporter driven by the HCV
neither of these scenarios has, to our knowledge, ever been IRES was completely resistant, as were uncapped or adenosine
observed. On the basis of polysome dynamics alone, an miRNA- (ApppG)–capped nonpolyadenylated mRNAs dependent on the
mediated 90% decrease in protein output with no change in the encephalomyocarditis virus (EMCV) IRES (29). Because initia-
polysome distribution of the target mRNA would require a 90% tion driven by the EMCV IRES requires all the canonical initia-
reduction in initiation frequency coupled with a balancing 90% tion factors except eIF4E, the cap-binding factor (31), and the
reduction in elongation rate, a somewhat implausible scenario. HCV IRES functions independently of eIFs 4A, 4B, 4E, and 4G
These considerations led to the suggestion, likewise made by (32), this result indicates that the repression mechanism affects
Olsen and Ambros, that initiation and elongation rates on the the function of eIF4E rather than any other factor.
repressed mRNA may be normal, and the strong reduction in These results are markedly similar to those in the second pa-
protein product yield could be the result of a specific cotrans- per, by Humphreys et al., who cotransfected HeLa cells with
lational degradation of the nascent protein chain (24). For this CXCR4 siRNA and various luciferase reporter mRNAs, each
reason, the question of whether any full-length protein product with four bulged target sites for this siRNA (33), the very same
can be detected following polysome runoff, or in pulse-labeling reporters as used by the Sharp group (8, 11). At 16 hours after
experiments, is important. In the Petersen et al. experiments, transfection, the degree of CXCR4 siRNA-dependent repression
pulse-labeling for 3 min failed to detect any product (12), even of a m7GpppG-capped polyadenylated mRNA was greater than
though the reporter had an N-terminal epitope tag to facilitate 80% or about 50% if the poly(A) tail was absent. In addition,
product detection, an observation that at face value is consistent 50% repression was seen with an ApppG-capped polyadenylated
with the cotranslational degradation hypothesis. mRNA that would be translated by the scanning ribosome mech-
However, there is an ingenious experiment that argues other- anism, and an ApppG-capped polyadenylated mRNA with an
wise. An mRNA encoding a protein with an N-terminal signal EMCV IRES, whereas no repression was detectable with non-
sequence for targeting the product to the endoplasmic reticulum polyadenylated versions of these RNAs or an ApppG-capped
(ER), and with three bulged sites for let-7a miRNA, was just as nonpolyadenylated mRNA with the CrPV IRES (33).
susceptible to repression by endogenous HeLa cell let-7a as was The sensitivity of the m7GpppG-capped mRNA to repression,
a luciferase reporter that encoded a cytoplasmic product (29). contrasted with the lack of response of the nonpolyadenylated
Because the ER-targeted nascent protein chain is unlikely to be mRNAs with the viral IRESs, is in complete agreement with Pil-
accessible to any cytoplasmic proteolytic system, this result lai et al. and points to the same conclusion—an inhibition of
argues against the cotranslational degradation hypothesis. It eIF4E function (29, 33). However, although the nonpolyadenylated
should be noted, however, that in other experiments these EMCV IRES construct was not repressed, adding a poly(A) tail

www.stke.org/cgi/content/full/2007/367/re1 Page 4
REVIEW

increased its translation efficiency (in the absence of the CXCR4 on overall reporter mRNA stability was detected in these
siRNA) by a factor of ~2.5, but rendered it susceptible to a 50% experiments, the methods used would not have detected
repression by this siRNA (33). In other words, the siRNA (al- changes in poly(A) tail length (33).
most) completely negated the stimulatory effect of the poly(A)
tail. This suggests that apart from any effect on eIF4E function, Can These Apparently Conflicting Results Be
the CXCR4 siRNA may also interfere with the “closed loop,” in Reconciled?
which poly(A) binding protein (PABP) bound to the poly(A) tail There are two, apparently unrelated, discrepancies between the
interacts with the eIF4G component of the eIF4F complex (Fig. data described above and the results of Petersen et al.: (i) the dif-
2) bound either to the 5′ end of the mRNA (strongly in the case ference in polysome distribution of the repressed mRNA, and
of m7GpppG-capped RNAs and more weakly with an ApppG- (ii) the issue of whether the viral IRESs are insensitive to repres-
cap) or to an internal site in the EMCV IRES (34, 35). This sion (12, 29, 33). Although technical details of the polysome
poly(A)-PABP–dependent “closed loop” enhances the efficiency analyses are not identical, there is no obvious difference that
of initiation of both scanning-dependent mRNAs and mRNAs de- might explain the disagreement. Both Pillai et al. and Petersen et
pendent on the EMCV IRES, but the HCV IRES is unaffected by al. used DNA transfections for these experiments, albeit over
the presence or absence of a poly(A) tail (36–39). different time periods (48 and 20 hours, respectively) (12, 29).
One can envisage two alternative ways by which the One clear difference is that Pillai et al. relied on endogenous
CXCR4 siRNA could disrupt the “closed loop,” either via a let-7a to effect the repression (29), whereas Petersen et al. used
disruption of the protein-protein and protein-RNA interactions exogenous CXCR4 siRNA (12), which might well have resulted
necessary to maintain the loop, or by simply promoting dead- in a higher intracellular concentration of the short RNA. This,
enylation of the mRNA. Although no notable effect of the siRNA together with the fact that Petersen et al. had six bulged target

Downloaded from stke.sciencemag.org on February 26, 2009


sites in their reporter as compared to just three in the reporter
A used in the other paper, may account for the higher repression
RNA helicase
(DEAD box) in their experiments, more than 95% as compared to 80 to 90%
in Pillai et al. One possibility is that at lower miRNA/mRNA
eIF4A target ratios and lower repression ratios, inhibition is exclusively
PABP
or largely at the initiation step, whereas with increasing severity
elF4GI 570-582 722-876 1211-1411 1560
of repression a secondary effect on elongation rate or processivity
130-160
elF4E
975-1065 eIF4A sets in.
Cap-binding
As for the issue of whether IRES-dependent mRNAs are
insensitive to repression, in this case there are differences in pro-
Sequence-independent
cedure that deserve consideration as a possible explanation for the
RNA-binding domain elF3 40S subunit discrepancy: DNA transfections of constructs that would produce
a capped dicistronic mRNA (12), as opposed to RNA transfec-
tions of monocistronic IRES-containing RNAs (29, 33). The
B capped dicistronic mRNA generated in the Petersen et al. proce-
dure would certainly bind the eIF4F complex (Fig. 2), and the
eIF4E m7GpppG AUG expected repression of the upstream cistron was indeed observed
40S (12). Is it possible that this repression of the upstream cistron,
4A however it may occur, could spread so as to also affect the down-
eIF
stream cistron? Unfortunately, this conjecture is undermined by
the results obtained by Pillai et al. with another type of dicistronic
eIF4G elF3 mRNA (29), in which downstream cistron expression was driven,
not by a natural viral IRES, but by tandem phage λ Box B sites in
the intercistronic spacer, and coexpression of an eIF4G-λ N pep-
BP UAA tide fusion protein, which would bind specifically to the Box B
PABP PA AA sites (Fig. 3C). In DNA transfection assays, the presence of three
AAA AA A
AAAAAAAAA bulged target sites for let-7a in the 3′-UTR of this dicistronic con-
struct resulted in 65% repression of upstream cistron expression
Fig. 2. Schematic diagram of (A) the eIF4F complex and (B) the with apparently no effect on the downstream cistron (29).
“closed loop” formed by the interaction of PABP bound to the
Different cell types and different miRNAs are unlikely to
poly(A) with the eIF4G component of the eIF4F complex bound to
explain the conflicting evidence implicating different mechanisms
the 5′-cap. Mammalian cells have two eIF4Gs encoded by different
genes. Various isoforms of the more abundant species, eIF4GI,
of miRNA action. Petersen et al. and Humphreys et al. used pre-
arise from initiation at different AUG codons by leaking scanning. cisely the same siRNA (CXCR4) and reporters with bulged target
The isoform shown, which is generally the most abundant, is the sites of precisely the same sequence (though the number of sites
second largest and lacks the 40 N-terminal amino acids of the differed), yet came to completely different conclusions (12, 33).
longest form. Apart from the direct interactions with eIF4A, eIF4E, Likewise, there appears to have been sufficient overlap of different
and PABP, eIF4G also interacts directly with eIF3, which itself cell types used by each group to be able to eliminate this as the
binds tightly to the 40S ribosomal subunit. Thus, there is an indi- explanation. Thus, there is no clear evidence pointing to one of the
rect eIF4G/40S subunit interaction bridged by eIF3, and this is two radically different outcomes being the right answer and the
thought to be instrumental in loading the 40S subunit on the mR- other wrong, and the collective published data show no sizable
NA and in facilitating ribosome scanning. majority vote in favor of either explanation. If this and other

www.stke.org/cgi/content/full/2007/367/re1 Page 5
REVIEW

recent reviews (40) appear to have a bias in favor of initiation A


being the regulated step, this is probably because there are numerous N–Ago/GW182
precedents for regulation of eukaryotic initiation by protein-
mRNA interactions at 3′-UTR sites, but no precedents for cotrans- Renilla luciferase
lational degradation of the nascent protein, or for high-frequency
premature ribosome drop-off. However, a more detached and
unbiased position would take the view that miRNAs may be able
AAAA
to mediate repression by either mechanism, but as these two out-
comes are so radically different, this would also imply that there 5 Box B hairpins
must be at least one unrecognized or uncontrolled variable that bind N-peptide
determines which of the two putative alternative mechanisms is Firefly luciferase as internal control
predominant in particular experimental situations.
There are actually some precedents, dating from over 20 years
B
ago, for unexpected polysome association of near-silent mRNAs.
Heat shock of Drosophila cells generally leads to a complete dis- RL
appearance of polysomes (due to inhibition of initiation) followed
by the appearance of polysomes specifically translating heat- ( )
shock mRNAs. However, in one report, some 30 to 50% of the
housekeeping mRNAs were still polysome- associated 1 hour af- FL RL
ter heat shock, even though the expression of the encoded

Downloaded from stke.sciencemag.org on February 26, 2009


proteins was reduced by at least 90% (41). Similarly, prolonged IRES
incubation of HeLa cells with amino acid analogs induces the
synthesis of stress proteins and shuts off synthesis of house-
N–elF4E*/elF4G*
keeping proteins, yet the mRNAs encoding these proteins remain C
largely polysome-associated (42). Inhibition of initiation was
thought to be the immediate response to such stress, but it was
suggested that an additional elongation defect might set in
somewhat later. More recent confocal microscopy work reveals FL RL
an interesting parallel with miRNA-mediated repression in
that these near-silent housekeeping mRNAs would be largely
2 to 6 Box B
localized in cytoplasmic bodies known as stress granules
(43, 44), whereas mRNAs subject to miRNA-mediated repres- Fig. 3. Examples of the IRES and tethered-function constructs
sion localize to another type of cytoplasmic foci known as described here. (A) A reporter construct with five phage λ Box B
P-bodies (see below). hairpins in its 3′-UTR is cotransfected with a construct designed
Of course, the big difference between these situations and to express a fusion protein consisting of phage λ N-peptide fused
miRNA-mediated repression is that the latter is selective for to a human or Drosophila Ago protein, or GW182 (and often with
(one or a few) particular mRNA species, whereas the stress con- an epitope tag spacer, which is not shown). The high-affinity in-
ditions affect the behavior of bulk mRNA. There are, however, teraction between the N-peptide moiety and Box B hairpins teth-
some examples in which a specific mRNA that appears to be ers the Ago or GW182 to the target mRNA. (B) Schematic dia-
inactive is nevertheless still found in polysomes: thymidine gram illustrating the type of monocistronic and dicistronic mR-
kinase mRNA in differentiating myoblasts (45), and nanos NAs with viral IRESs discussed here. (C) A dicistronic mRNA in
which the downstream cistron expression is dependent on an ar-
mRNA in Drosophila embryos (46). The polysomes with
tificial IRES system consisting of two or more phage λ Box B
repressed nanos mRNA were sensitive to EDTA, they decreased
hairpins. The DNA construct is cotransfected with constructs de-
in size on prolonged incubation of the embryo extract with signed to express a fusion protein consisting of phage λ N-pep-
puromycin, and polysome runoff resulted in release of most of tide fused to mammalian eIF4E or eIF4G mutants (eIF4E*,
the nanos mRNA, yet no Nanos protein product could be detected eIF4G*) unable to function in cap-dependent initiation. The
(46). These results are similar to the original observations on the eIF4G mutant is unable to interact with eIF4E but retains the
status of repressed lin-14 and lin-28 mRNAs in C. elegans functionally important interactions with eIF3 and eIF4A (Fig. 2).
L2/L3 larvae (24, 25), and to the results of Petersen et al. (12). The eIF4E mutant cannot bind to 5′-caps, but retains the poten-
tial for interaction with wild-type endogenous eIF4G, and thus the
miRNAs Can Also Promote Target mRNA Degradation binding of the N-peptide/eIF4E* fusion protein to the Box B hair-
In almost all published studies, there is at least some miRNA- pins recruits endogenous eIF4G to the vicinity of the down-
dependent decrease in mRNA abundance, which has usually stream cistron initiation site. Relevant references cited in this
been ignored, as it is often relatively small compared with the work are as follows: (A) (19, 20, 29); (B) (12, 26, 29); (C) (29).
inhibition of expression of the encoded protein. For example,
Petersen et al. observed a greater-than-95% decrease in reporter tion as an important aspect of miRNA-mediated repression of
expression, but only a 50% decrease in mRNA abundance (12). gene expression. Microarray analyses of HeLa cell mRNAs
Nevertheless, an explanation for this 50% decrease in mRNA showed that expression of the brain-specific miR-124 or the
abundance is called for. muscle-specific miR-1 decreased the abundance of different
Some recent publications report larger miRNA-dependent subsets of 100 to 200 mRNAs in each case, specifically mRNAs
decreases in mRNA abundance, and emphasize mRNA degrada- that are of low abundance in brain and muscle, respectively (47).

www.stke.org/cgi/content/full/2007/367/re1 Page 6
REVIEW

In addition, miRNA-dependent degradation of mRNAs that have suggesting almost no effect on translation efficiency per se; and
3′-UTR AU-rich destabilizing elements has been reported, even the Vha68-1 3′-UTR/miR-9b pair gave results intermediate be-
though it is not the “seed” sequence of this miRNA (miR-16) tween these two extremes (20).
that is complementary to the AU-rich sequence (48). Even more A consequence of deadenylation is disruption of the “closed
surprising is a recent report that in C. elegans larvae, lin-4 and loop” (Fig. 2), resulting in a severe reduction in translation effi-
let-7 miRNAs promoted extensive degradation of lin-14, lin-28, ciency, which will fall effectively to zero after the subsequent
and lin-41 target mRNAs (49), notwithstanding the previous evi- step of decapping. How much the low translation efficiency of
dence for repression of translation of these mRNAs (24, 25). this capped deadenylated species contributes to the true repres-
However, in none of these cases was there evidence that degra- sion of translation that is observed will depend on the lifetime
dation occurred by the endonucleolytic mechanism characteristic of this transient decay intermediate and its steady-state concen-
of RNAi by perfectly complementary siRNAs (40). tration relative to that of the polyadenylated species. These
Three recent papers, two concerned with mammalian sys- considerations are well illustrated by the three Drosophila
tems and the other with Drosophila cells, also demonstrate mRNAs mentioned above, where RNAi knockdown of decap-
miRNA-mediated acceleration of target mRNA degradation, not ping enzymes caused the target mRNA abundance to increase
by an siRNA-like mechanism of endonucleolytic cleavage, but (by trapping the decay intermediates as capped deadenylated
rather, through the normal pathway of deadenylation, followed transcripts), but the increase in reporter protein output was
by decapping and subsequent degradation of the body of the smaller, so that the degree of true translational repression actu-
mRNA by 5′→3′ exonuclease activity (18, 20, 50). The only ally increased, in the sense that the ratio of protein output to the
difference from the normal pathway is that the miRNP bound to amount of mRNA was reduced (20). However, because siRNA-
its 3′-UTR target sites can apparently act as a barrier to the mediated depletion of deadenylating enzymes also increased

Downloaded from stke.sciencemag.org on February 26, 2009


5′→3′ exonuclease, leading to accumulation of deadenylated target mRNA concentrations (to a lesser extent), but this time
decay intermediates with 3′-end sequences (18, 49). with very little change in the degree of actual translational
Thus, in Drosophila cells, target mRNA concentrations repression (the ratio of protein output to mRNA abundance), it
could be restored by RNAi knockdown not only of Ago1 but al- seems likely that under normal conditions, translational repres-
so of the deadenylating (Not1 and Ccr4) or decapping enzymes sion is occurring mainly on polyadenylated mRNAs, with com-
(Dcp1/2), and in the last case deadenylated capped mRNAs ac- paratively little contribution from the low translation efficiency
cumulated (20). In mammalian cells with a target gene under of the capped deadenylated mRNA decay intermediates. (The
control of the c-fos promoter (to allow a transient transcriptional situation may be somewhat different in RNA transfection
pulse), an acceleration of deadenylation (and subsequent decay assays, where there is no ongoing production of fully
of the mRNA body) dependent on miR-125b expression was polyadenylated mRNAs.)
seen even with a reporter that had just a single bulged 3′-UTR In mammalian cells expressing (from a CMV promoter) a
target site for this miRNA (50). The accelerated deadenylation polyadenylated luciferase reporter mRNA with six bulged target
was not dependent on translation of the mRNA, because it was sites for miR-125b, coexpression of this miRNA reduced mRNA
not substantially affected by insertion of a stable hairpin in the abundance by about 70%, and luciferase protein expression by
5′-UTR. Nor did premature decapping occur on the mRNAs more than 90%, indicative of a 65% decrease in the actual trans-
that had not yet been completely deadenylated (50). lation efficiency (50). When the poly(A) tail was replaced by the
Similarly, in zebrafish embryos, the expression of the miR- 3′-terminal stem-loop of histone H1.3, miR-125b coexpression
430 family of microRNAs, which starts at ~5 hours after fertil- had little effect on mRNA abundance (just a 10% decrease), but
ization, led to deadenylation and subsequent degradation of sev- reduced translation efficiency by about 70%. The close corre-
eral hundred maternal mRNAs with predicted bulged target spondence between the reduction in true translation efficiency in
sites (and injected reporter mRNAs with such sites) at the stage these two experiments likewise suggests that the poor translata-
when zygotic transcription is activated. This serves to clear the bility of capped deadenylated decay intermediates, which would
maternal mRNAs rapidly from the embryo and thus makes the be present only in the former case, contributes little to the
translation machinery available for the new transcripts. In mu- observed translational repression. The issue of whether the
tant embryos that do not express the miR-430 microRNAs, inhibition was exerted at initiation of translation or at some
deadenylation and degradation of the maternal transcripts still subsequent step in protein synthesis was not addressed in this
occurred but were delayed, which led to abnormalities in brain study, but if future work indicates inhibition of initiation, it is
development (51). worth bearing in mind that, despite the absence of a poly(A) tail,
Thus, mRNA degradation and inhibition of translation ap- there is evidence that a “closed loop” (Fig. 2) can form with
parently both contribute to the overall repression of gene ex- histone mRNAs and that this stimulates their translation (52, 53).
pression by miRNAs, but the relative importance of each mech-
anism may vary according to the cell type and the stability or miRNAs and Their mRNA Targets Accumulate in
configuration of the particular miRNA-mRNA pair (18, 20, 50). P-Bodies
This dual explanation for decreased expression is well illustrat- Another important development has been the discovery that
ed by the data for reporters with the 3′-UTRs of three different Argonaute proteins, miRNAs, and their target mRNAs accumu-
Drosophila genes: With the nerfin 3′-UTR, overall reporter pro- late in cytoplasmic foci usually known as P-bodies, or process-
tein output was reduced 90% by miR-9b expression, but mRNA ing bodies, which are detected by confocal microscopy and
levels were reduced by only 20%, indicative of an effect pre- immunostaining or the use of fusions between candidate P-body
dominantly on translation eff iciency; with the CG10011 proteins and a fluorescent protein (29, 54, 55). The protein
3′-UTR, protein output and mRNA abundance were both composition of these foci always includes enzymes that are
reduced by the same extent (80%) by miR-12 expression, important in the normal pathway of mRNA degradation, but is

www.stke.org/cgi/content/full/2007/367/re1 Page 7
REVIEW

more complex in higher eukaryotes than in yeast, where it that after storage and sorting in stress granules, some mRNAs
includes the Dcp1/Dcp2 decapping complex, Xrn1 5′-3′ exonu- may pass to P-bodies for degradation.
clease, most of the Lsm proteins (Lsms 1 through 7, a family of Entry of an mRNA into Dcp1/Dcp2 foci does not inevitably
related RNA-binding proteins necessary for mRNA decay), the lead to its degradation, as shown by the fact that glucose starva-
Dhh1p putative RNA helicase (known as RCK or p54 in verte- tion of yeast, which results in a global inhibition of initiation,
brates), and Pat1p, but not ribosomes, nor any of the eight trans- drives mRNAs into P-bodies, but on refeeding they exit the foci
lation initiation factors examined, nor components of the exo- and become (re)assimilated into active polysomes (58, 69).
some complex of 3′→5′ exonucleases (56–58). Although Pat1p Reversibility of miRNA-dependent repression and P-body
homologs have not been tested, P-bodies in higher eukaryotes localization has been demonstrated for the cationic amino acid
seem to have the same proteins plus many additional compo- transporter (CAT-1) mRNA, which has three or four [depending
nents, most of them absent from the yeast proteome, and on which poly(A) addition site was used] bulged 3′-UTR sites
because budding yeast neither expresses siRNAs or miRNAs nor for interaction with miR-122 (30). In Huh7 cells, the endoge-
shows any response to ~21-residue RNAs, at least some of these nous miR-122 reduced CAT-1 protein levels by about 65%
additional components in higher eukaryotes could play a role in under fed conditions (as judged by the results of introducing an
the mechanisms of siRNA- and miRNA-mediated mRNA silenc- antisense oligonucleotide to this miRNA), and both the CAT-1
ing. Interesting potential candidates include the following mRNA and the miR-122 could be detected in P-bodies. On
proteins (59–62): GW182, a putative RNA-binding protein with amino acid starvation there was a rapid increase in the amount
glycine-tryptophan repeats; eIF4E (but no other translation initi- of CAT-1 protein, with no immediate change in mRNA abun-
ation factors nor PABP nor ribosomes); and the eIF4E-binding dance; rather, the preexisting CAT-1 mRNA left the P-bodies
protein, 4E-T (but not 4E-BP1, another eIF4E binding protein). and was mobilized into polysomes. An unexpected additional

Downloaded from stke.sciencemag.org on February 26, 2009


The Drosophila 4E-T homolog, known as Cup, has been impli- finding was that both the activation of translation and the mobi-
cated in the repression of specific maternal mRNAs (63–65), lization from P-bodies required a particular AU-rich element
whereas 4E-BP1 interaction with eIF4E results in global inhibi- (ARE) in the 3′-UTR and the presence of the ARE-binding pro-
tion of initiation (66). tein, HuR (30). Together, these observations conf irm that
P-bodies are highly motile within the cell cytoplasm (44) miRNA-mediated repression and P-body localization of the
and fluctuate in size and number. In situations where P-bodies target mRNA are potentially reversible processes. P-bodies can
seem to disappear as visible foci, it is not clear whether their be regarded as a repository for untranslated mRNA, some of
components become completely dispersed in the cytoplasm, or which may subsequently be degraded, whereas some may be
whether “mini” P-bodies persist that are undetectable by confo- returned to translation. An unresolved question is whether the
cal microscopy. P-bodies not only contain (untranslated) mRNA translational activation of CAT-1 mRNA is accompanied by
and especially mRNA decay intermediates, but they require this complete dissociation of the miRNP from the target mRNA, or
RNA for their integrity (57, 60). Thus, the foci disappear in whether it is just a case of the repressive action being overrid-
yeast mutants with defective deadenylation, which would den in some way without dissociation.
reduce the level of decay intermediates, and increase in size and It was estimated that some 20% of let-7a miRNA and 20%
number in mutants defective in downstream steps of the decay of its target mRNA in HeLa cells were localized in foci, under
process, such as mutants in the Dcp1/Dcp2 decapping complex conditions in which the target was repressed by at least 80%
or the Xrn1 5′→3′ exonuclease (56, 57). Similar results are (29). The results of biochemical fractionation of cell extracts
seen in mammalian cells following RNAi knockdown of mRNA prepared by lysis with digitonin suggest that the repression of
degradation enzymes (60, 61), and in addition, P-bodies the rest of the target mRNA occurred in “mini” P-bodies smaller
decrease following depletion or knockdown of GW182, 4E-T, than the threshold size detectable by confocal microscopy,
Lsm1, and the RCK/p54 helicase (61, 67). Inhibition of transla- rather than uniformly and completely dispersed throughout the
tion elongation (e.g., by cycloheximide) causes the foci to dis- cytoplasm. Centrifugation of these extracts at 14,000g pelleted
appear, whereas inhibition of initiation results in an increase in almost all the target mRNA (but not the tubulin mRNA exam-
size and number (56–58, 60, 61), suggesting that an mRNA has ined as an unrepressed control), most of the let-7a miRNA (in
to be cleared of ribosomes before it can localize to P-bodies. the case of HeLa cells), and all of the endogenous miR-122
Thus, the P-body localization of the mRNA targets of miRNA- (Huh7 cells), together with all of the cellular complement of the
mediated repression can be taken as additional, though circum- P-body marker proteins, Xrn1 and Dcp1 (29, 30).
stantial, evidence that it is initiation that is the inhibited step. It seems intuitive that it is the inhibition of translation rather
Irrespective of whether repression was effected by endoge- than the mere binding of the miRNP to the target mRNA that
nous let-7a miRNA or exogenous CXCR4 siRNA, a fraction of drives the repressed mRNA into P-bodies. Thus, it would be
the target mRNA was localized to foci resembling P-bodies (29, reasonable to assume that in the experiments where mRNAs
54). However, Pillai et al. emphasize that the foci in which the with the EMCV, HCV, or CrPV IRESs were resistant to repres-
repressed mRNA and let-7 miRNA were localized did not sion (29, 33), an miRNP-mRNA complex still forms on these
always coincide precisely with the Dcp-containing foci (29): mRNAs, yet presumably they do not get localized to P-bodies.
67% were perfectly overlapping with Dcp-1 foci, and 37% were It would be intriguing to know what happens as regards P-body
described as being adjacent. Furthermore, there were a few Dcp accumulation with a dicistronic mRNA (Fig. 3C), in which the
foci apparently lacking the target mRNA, and vice versa, as upstream cistron is sensitive to miRNA-mediated repression but
though there might be two subtypes of foci that might fuse the downstream cistron is insensitive (29).
occasionally or exchange mRNA. In a similar vein, adjacent Another case where it seems most unlikely that the target
location and possible fusion between P-bodies and stress gran- RNA enters P-bodies is the interaction of miR-122 with HCV
ules has also been noted (43, 44, 68), leading to the suggestion RNA, which occurred at a 5′-proximal site upstream of the

www.stke.org/cgi/content/full/2007/367/re1 Page 8
REVIEW

IRES and was necessary for the replication of the viral RNA, translation was specifically repressed at the initiation step by up
without having any direct effect on its translation (70). Here to 80%, dependent on the number of Box B sites, but indepen-
again, there is no reason to suppose that an miRNP complex dent of their position in the 3′-UTR, with no appreciable de-
with Ago proteins does not form on the viral RNA, yet it seems crease in mRNA abundance (19, 29). Curiously, although re-
most unlikely that this results in the miRNP/viral RNA complex pression promoted by tethering Ago bypasses the requirement
accumulating in P-bodies. for an miRNA, repression and P-body localization did not occur
with tethered Ago PAZ domain mutants that are defective in
siRNA-Mediated Knockdown and Tethered Function binding the miRNA/mRNA complex (67).
Approaches to Identify the Essential Mediators of Tethering GW182 to an mRNA in the same way mimicked
miRNA-Dependent Repression of Translation miRNA-mediated repression in Drosophila cells, causing a
Approaches toward elucidating the mechanism of repression in- 75% decrease in the abundance of the reporter mRNA and a
clude (i) identifying which proteins interact with Argonaute pro- greater than 90% overall reduction of luciferase output, imply-
teins, (ii) testing whether miRNA-mediated repression is relieved ing a 75% inhibition of translation per se (20). The same effects
by siRNA-mediated knockdown or depletion of these and other of tethered GW182 were seen even in cells depleted of Ago1.
likely candidate proteins, and (iii) tethered function assays (Fig. 3). Thus, tethering the relevant Argonaute protein can bypass the
Proteins that interact with mammalian Ago1 or Ago2 pro- miRNA requirement for repression, and tethering GW182
teins in an RNase-insensitive manner, and colocalize in P-bodies, bypasses the Ago requirement, at least in Drosophila cells,
include the Dcp1-Dcp2 decapping complex, GW182, and suggesting that GW182 acts downstream of Ago proteins.
RCK/p54 helicase, which is the vertebrate homolog of yeast As for p54, tethering it to the 3′-UTR of a reporter mRNA
Dhh1p (54, 67, 71). The interactions with Dcp1/2 and GW182 specifically repressed reporter translation in Xenopus oocytes,

Downloaded from stke.sciencemag.org on February 26, 2009


still occurred with Ago mutants deficient in RNA binding that implying that p54 plays an important role in the represssion of
did not localize to P-bodies (54, 67), and the interaction with maternal mRNA in these oocytes (73, 74). The Drosophila p54
p54 still occurred in cells depleted of Lsm1 (a protein found in homolog, Me31B, has also been implicated in the repression of
P-bodies and implicated in RNA processing), which dispersed certain maternal mRNAs in early embryos, and in this capacity
the foci (71). Again, it is not clear whether these interacting it appears to act as a corepressor working in conjunction with
proteins were completely dispersed in these situations. The Cup, the Drosophila homolog of mammalian 4E-T (75, 76). It
Drosophila GW182 homolog binds to and localizes with Ago1, is not yet known if tethering 4E-T/Cup to the 3′-UTR also
but not Ago2 (20), which functions mainly or exclusively in the represses translation, and, if so, whether it or p54 acts down-
siRNA-mediated endonucleolytic pathway. stream of GW182 or vice versa.
When Drosophila cells were depleted of these various pro-
teins, relief of miRNA-mediated repression was seen with Recapitulation of miRNA-Mediated Repression in Vitro
knockdown of Ago1, GW182, and Dcp1/Dcp2, in descending Many of the uncertainties about the exact mechanism of transla-
order of efficacy (20, 72). Depletion of Xrn1 had no effect, nor tional repression might be resolved if repression could be reca-
did Ago2 knockdown, though this did relieve siRNA-mediated pitulated in an in vitro system, as has been achieved for siRNA-
endonucleolytic cleavage of mRNA with a perfectly comple- mediated endonucleolytic cleavage (77, 78). Until recently, no
mentary target site (72). In mammalian cells, repression of re- such in vitro recapitulation of miRNA-mediated repression had
porters with either four or six bulged target sites for CXCR4 been reported, although we presumed that it had been attempted
siRNA was likewise relieved by GW182 depletion and, to a in many laboratories without success. However, success has
slightly lesser extent, by knockdown of p54 helicase or Dcp2, recently been achieved with the same siRNA (CXCR4) and a
but not by Xrn1 knockdown (67, 71). Depletion of Lsm1, which luciferase mRNA with the same bulged target sites as used by
caused dispersal of P-bodies, had no effect on the degree of re- Petersen et al. (12) and Humphreys et al. (33). The essential
pression, implying that the usual localization in visible foci is a trick was to pre-anneal the miRNA to the target mRNA before
consequence of repression rather than a prerequisite for it (71). addition to the rabbit reticulocyte lysate (RRL) system (79).
In the case of a reporter with a single site perfectly complemen- Although this requirement for pre-annealing means that the
tary to the introduced siRNA, depletion of GW182 again gave miRNP-mRNA complex was not assembled by the normal route
some relief, though not as much as Ago2 knockdown, and that is thought to occur in intact cells, the outcome had all the
Dcp1/2 or Xrn1 depletion had no effect (67, 71). hallmarks of miRNA-mediated regulation established in vivo:
In view of the indications that miRNA-mediated repression in increased repression with increasing number of target sites; a
some way affects eIF4E function (29, 33), it would be interesting strict requirement for a 5′-phosphate on the miRNA; require-
to test the outcome of 4E-T knockdown. Another candidate ment for perfectly complementary contiguous base-pairing of
worth testing would be the higher-eukaryote homolog of yeast residues 2 to 8 of the miRNA, coupled with a much less strin-
Pat1p, which has been shown to act as a global translational re- gent requirement for contiguous base-pairing of the 3′ end; no
pressor together with Dhh1p (the yeast p54 homolog), in an ad- repression if the 3′-end of the 21-nt miRNA was extended by 10
ditive rather than overlapping way (69). In strains deleted for residues perfectly complementary to the target mRNA; and no
both, the normal response to glucose starvation, which includes repression with a 2′-O-methyl derivative of the 21-nt mRNA.
the breakdown of polysomes, cessation of translation, and mobi- Although the commercial RRL system used is one that appears
lization of all mRNAs into P-bodies, no longer occurred (69). to have been optimized for translation of uncapped mRNA,
Finally, tethered function assays have shown that coexpres- repression was only seen if the mRNA was both capped and had
sion of Ago-λ N-peptide fusions and a reporter with tandem a standard-length poly(A) tail (200 A residues), but uncapped
phage λ Box B sequences in its 3′-UTR (Fig. 3A) caused the re- mRNAs were repressed if the tail was extended to unphysiolog-
porter to behave as if it were repressed by microRNAs. Its ical lengths (2000 residues).

www.stke.org/cgi/content/full/2007/367/re1 Page 9
REVIEW

Maximal repression (70% reduction) of luciferase output A Accelerated


was seen at short incubation times (10 min), and the time- deadenylation
course data implied that the CXCR4 siRNA increased the lag
time before the luciferase activity first appeared, after which the Inhibition
actual rate of increase was not very different. Although these of initiation
miRNP
kinetics could imply a delay in the first initiation events, or
alternatively a slower rate of elongation, it could also be that the m7GpppG AUG UAA AAAAAAAA
repression mechanism is rather labile over time, a suggestion
which would be consistent with the finding that repression by A
CXCR4 siRNA was more sensitive to freeze-thawing of the Defective B
RRL than was the basal translation activity (79). Although the elongation
data do not allow an unambiguous identification of which step
of translation is being inhibited, the requirement for a cap and a
poly(A) tail strongly suggests that it is initiation rather than
elongation that is affected.
If the reporter mRNA had a single 3′-UTR site perfectly A. Cotranslational degradation of nascent protein
B. Premature ribosome drop-off
complementary to the CXCR4 siRNA, it appeared to be exten- C. Reduced elongation rate (and initiation frequency)
sively degraded in this system, implying that despite the
abnormal pathway, a competent RISC with an active Ago2 can
assemble in RRL, although confirmation of this requires a B

Downloaded from stke.sciencemag.org on February 26, 2009


demonstration that the reporter was cleaved in the expected site.
AAA
A Possible Model? Pat1
AA
AA
elF4E AA
We propose a speculative model to account for the available Ccr4 A
findings (Fig. 4). Figure 4A depicts all the different ways by

AA
Not1

AA
which miRNAs have been proposed to regulate their target 4E-T p54

A
AAA
mRNAs, other than the rare cases where metazoan miRNAs

AA
have near-perfect complementarity with an mRNA target site GW182 Dcp1/2
and therefore promote endonucleolytic cleavage (7). There are
two reasons why we are unable to enlarge upon the possible
mechanisms of perturbation of elongation that result in the Ago
repressed mRNA being found in polysomes of approximately
the same size as when it is unrepressed. First, it is not yet at all
clear whether this is due to cotranslational cleavage of the
nascent protein chain, or to premature ribosome drop-off Fig. 4. Ultraspeculative model for how miRNAs may be able to
(coupled with a compensating increase in initiation frequency), regulate gene expression at the posttranscriptional level. (A) A
or to a reduced elongation rate coupled with a similar decrease schematic depiction of the different mechanisms proposed for
the regulation of target mRNA function by miRNAs. Three
in the rate of initiation. Second, none of the components of
alternative ways in which miRNA-mediated repression might
miRNPs, and none of the proteins that have been found to inter-
perturb translation elongation are listed, with the green sphere
act with Ago proteins, is thought to interact with, or have any representing an unknown ribosome-associated protease
impact upon an elongating ribosome.. degrading the nascent protein chain cotranslationally. (B) Dia-
However, we are able to elaborate the model in respect of gram showing how the binding of an Ago protein to the site of
miRNA-mediated regulation of translation initiation and miRNA/mRNA interaction might lead to inhibition of translation
miRNA-dependent acceleration of mRNA degradation (Fig. initiation and an enhanced rate of deadenylation. (See text for
4B). In this scheme, binding of an Ago protein at the site of an explanation.) For convenience, the diagram depicts an
miRNA interaction with its bulged target sequence leads to mRNA with only a single target site for the miRNA. With multi-
recruitment of GW182, RCK/p54, the Dcp1/Dcp2 decapping ple sites there is the possibility that protein interactions at one
complex, 4E-T, and the higher eukaryote homolog of yeast site could be directed mainly toward inhibition of initiation, and
Pat1p (although there is as yet no evidence for or against those at another site toward deadenylation, or even toward
recruitment of these last two components). With the assis- aberrant elongation.
tance of p54, 4E-T interacts with eIF4E bound to the 5′-cap,
to form an inhibitory “closed loop” similar to the postulated can it explain how and why translation dependent on the
mechanism for repression of maternal mRNAs in Drosophila HCV and CrPV IRESs is susceptible to repression in some
embryos by the combined action of Cup and Me31B, the experiments (12). But apart from these problems, the model
Drosophila homologs of 4E-T and p54, respectively (63–65, is consistent with the other results discussed here, subject to
75). An additional assumption is that the bound Ago protein the minor caveat that the weak repression seen with ApppG-
recruits, either directly or indirectly, the Not1 and Ccr4 dead- capped polyadenylated mRNAs (with or without an EMCV
enylation factors, which results in accelerated shortening of IRES) in the RNA transfection assays of Humphreys et al.
the poly(A) tail and consequently accelerated mRNA degra- would have to be explained as a secondary consequence of
dation. This model cannot explain why repressed mRNAs and deadenylation rather than a direct result of any inhibition of
miRNAs sometimes end up in polysomes (12, 24, 25), nor initiation (33).

www.stke.org/cgi/content/full/2007/367/re1 Page 10
REVIEW

Concluding Remarks an EDTA-sensitive manner (81). This polysomal association was


A comment found with increasing frequency in the literature is dependent on base-pairing between the miRNA and mRNA,
that there may well be more than one mechanism by which miRNAs because the let-7a miRNA was found at the top of the gradient if
repress expression of their target genes. There is clear evidence the cells had been transfected with a 2′-O-methyl oligonu-
for this in respect to deadenylation-dependent degradation and cleotide antisense to this miRNA. Moreover, the polysomes were
true translational repression, and it does not stretch credulity too dynamic by the criteria that the miRNAs were found nearer the
far to imagine that the miRNA/mRNA interaction could lead to top of the gradient if the cells had been incubated with
both these outcomes. What does defy the imagination, on the puromycin or with initiation inhibitors before lysis (81).
other hand, is how the same miRNA-mRNA target pairs can In the other study, HeLa cells were transfected with a DNA
show inhibition of translation initiation in one system, but inhi- construct with a thymidine kinase promoter that had the Renilla
bition at some step after initiation in another. We embarked on luciferase coding region upstream of the C. elegans lin-41
this Review with the hope of finding an explanation for this ap- 3′-UTR, which has two validated target sites for let-7a miRNA
parent contradiction. We have failed in this aim, but some point- (82). As compared with a control lacking these two sites,
ers to future directions have emerged on the route to this failure. luciferase expression was reduced by 90%, with a decrease in
First, more attention should be paid than hitherto to the target mRNA abundance of 30%, implying an ~80% reduction
poly(A) tail length of the target mRNA. Moreover, target in true translation efficiency. About half of the target mRNA,
mRNA abundance should in future be assessed by methods as well as half the endogenous let-7a miRNA and AGO pro-
(e.g., Northern blotting) that, unlike RNase protection or teins, cosedimented with large polysomes (with the other half
reverse transcription–polymerase chain reaction amplification found at the top of the gradients), but they sedimented nearer
(RT-PCR) of a short internal segment, can distinguish full- the top of the gradient following incubation with puromycin or

Downloaded from stke.sciencemag.org on February 26, 2009


length mRNA from degradation intermediates. Second, it would under conditions of inhibited initiation (82). These results were
be interesting to know whether depletion of critical P-body taken as evidence that repression was occurring at some stage
components such as GW182 and RCK/p54 also relieves the re- after initiation, and the authors specifically favor the cotransla-
pression in those systems that lead to polysomal accumulation tional nascent protein degradation hypothesis, because, like
of the repressed mRNA. Third, it would be useful if those who Petersen et al. (12), they were unable to detect any nascent
find that target mRNAs with an HCV, CrPV, or EMCV IRES peptide product under repressed conditions, even though the
are insensitive to repression in RNA transfection assays were to reporter had an N-terminal epitope tag to facilitate detection of
confirm this result with dicistronic mRNAs, and also in DNA such peptide products.
transfections of mono- and dicistronic constructs. For this pur-
pose, it would also be worthwhile replacing the HCV IRES with
References and Notes
the phylogenetically related, but much more potent, IRES from 1. B. P. Lewis, C. B. Burge, D. P. Bartel, Conserved seed pairing, often
classical swine fever virus (80) as a counter to the argument flanked by adenosines, indicates that thousands of human genes are
(40) that translation dependent on some viral IRESs may be microRNA targets. Cell 120, 15–20 (2005).
2. J. Lu, G. Getz, E. A. Miska, E. Alvarez-Saavedra, J. Lamb, D. Peck, A.
unresponsive to miRNAs simply because these IRESs are Sweet-Cordero, B. L. Ebert, R. H. Mak, A. A. Ferrando, J. R. Downing, T.
inefficient. Fourth, because initiation on all the viral IRESs Jacks, H. R. Horvitz, T. R. Golub, MicroRNA expression profiles classify
used to date is not only independent of eIF4E but also does not human cancers. Nature 435, 834–838 (2005).
3. L. He, J. M. Thomson, M. T. Hemann, E. Hernando-Monge, D. Mu, S.
involve any ribosome scanning, it seems important to extend the Goodson, S. Powers, C. Cordon-Cardo, S. W. Lowe, G. J. Hannon, S. M.
range of IRESs studied to those that require eIF4E and those Hammond, A microRNA polycistron as a potential human oncogene.
where there is ribosomal scanning, so as to confirm that it is Nature 435, 828–833 (2005).
4. I. Alvarez-Garcia, E. A. Miska, MicroRNA functions in animal develop-
eIF4E function rather than scanning that is inhibited. Fifth, ment and human disease. Development 132, 4653–4662 (2005).
further use of depletions can be expected to identify more can- 5. V. Ambros, B. Bartel, D. P. Bartel, C. B. Burge, J. C. Carrington, X. Chen,
didate proteins required for repression, and tethered function G. Dreyfuss, S. R. Eddy, S. Griffiths-Jones, M. Marshall, M. Matzke, G.
assays coupled with knockdowns may reveal the hierarchy in Ruvkun, T. Tuschl, A uniform system for microRNA annotation. RNA 9,
277–279 (2003).
which these function. Sixth, further development of the cell-free 6. Y. Tomari, P. D. Zamore, Perspective: Machines for RNAi. Genes Dev.
systems (79) can be expected to illuminate the proximal mecha- 19, 517–529 (2005).
nism of repression, though it would be somewhat surprising if 7. S. Yekta, I. H. Shih, D. P. Bartel, MicroRNA-directed cleavage of HOXB8
mRNA. Science 304, 594–596 (2004).
such systems were able to shed light on the mechanisms of 8. J. G. Doench, C. P. Petersen, P. A. Sharp, siRNAs can function as
deadenylation and target mRNA accumulation in P-bodies. miRNAs. Genes Dev. 17, 438–442 (2003).
Finally, in order to facilitate reconciliation of the biochemical 9. M. Kiriakidou, P. T. Nelson, A. Kouranov, P. Fitziev, C. Bouyioukos, Z. Moure-
latos, A. Hatzigeorgiou, A combined computational-experimental approach
fractionation data with the confocal microscopy results, it predicts human microRNA targets. Genes Dev. 18, 1165–1178 (2004).
would be extremely useful to know what happens to P-bodies, 10. J. Brennecke, A. Stark, R. B. Russell, S. M. Cohen, Principles of
and where they and their constituent proteins partition, in the pro- microRNA-target recognition. PLoS Biol. 3, e85 (2005).
11. J. G. Doench, P. A. Sharp, Specificity of microRNA target selection in
cedures used by different laboratories to prepare cytoplasmic ex- translational repression. Genes Dev. 18, 504–511 (2004).
tracts for fractionation by sucrose density gradient centrifugation. 12. C. P. Petersen, M.-E. Bordeleau, J. Pelletier, P. A. Sharp, Short RNAs re-
Note added in proof. There have been two very recent publi- press translation after initiation in mammalian cells. Mol. Cell 21,
cations on these controversial issues. Both agree that repression 533–542 (2006).
13. D. Didiano, O. Hobert, Perfect seed pairing is not a generally reliable
occurs while the target mRNA is polysome-associated, implying predictor for miRNA-target interactions. Nat. Struct. Mol. Biol. 13,
that the repression must be exerted at some stage after initiation. 849–851 (2006).
In sucrose gradient analyses of HeLa cell polysomes, virtually 14. J. Liu, M. A. Carmell, F. V. Rivas, C. G. Marsden, J. M. Thomson, J. J.
Song, S. M. Hammond, L. Joshua-Tor, G. J. Hannon, Argonaute2 is the
the whole cellular complement of three representative miRNAs catalytic engine of mammalian RNAi. Science 305, 1437–1441 (2004).
(let-7a, miR-16, and miR-21) cosedimented with polysomes in 15. G. Meister, M. Landthaler, A. Patkaniowska, Y. Dorsett, G. Teng, T.

www.stke.org/cgi/content/full/2007/367/re1 Page 11
REVIEW

Tuschl, Human Argonaute2 mediates RNA cleavage targeted by driven translation but not for X-mediated stimulation of hepatitis C virus
miRNAs and siRNAs. Mol. Cell 15, 185–197 (2004). translation. Mol. Cell. Biol. 21, 4097–4109 (2001).
16. J. S. Parker, S. M. Roe, D. Barford, Crystal structure of a PIWI protein 40. M. A. Valencia-Sanchez, J. Liu, G. J. Hannon, R. Parker, Control of
suggests mechanisms for siRNA recognition and slicer activity. EMBO translation and mRNA degradation by miRNAs and siRNAs. Genes
J. 23, 4727–4737 (2004). Dev. 20, 515–524 (2006).
17. J. B. Ma, K. Ye, D. J. Patel, Structural basis for overhang-specific small inter- 41. D. G. Ballinger, M. L. Pardue, The control of protein synthesis during
fering RNA recognition by the PAZ domain. Nature 429, 318–322 (2004). heat shock in Drosophila cells involves altered polypeptide elongation
18. D. Schmitter, J. Filkowski, A. Sewer, R. Pillai, E. Oakeley, M. Zavolan, P. rates. Cell 33, 103–113 (1983).
Svoboda, W. Filipowicz, Effects of Dicer and Argonaute down-regulation 42. G. P. Thomas, M. B. Mathews, Alterations of transcription and transla-
on mRNA levels in human HEK293 cells. Nucleic Acids Res. 34, tion in HeLa cells exposed to amino acid analogs. Mol. Cell. Biol. 4,
4801–4815 (2006). 1063–1072 (1984).
19. R. S. Pillai, C. G. Artus, W. Filipowicz, Tethering of human Ago proteins 43. N. Kedersha, G. Stoecklin, M. Ayodele, P. Yacono, J. Lykke-Andersen,
to mRNA mimics the miRNA-mediated repression of protein synthesis. M. J. Fitzler, D. Scheuner, R. J. Kaufman, D. E. Golan, P. Anderson,
RNA 10, 1518–1525 (2004). Stress granules and processing bodies are dynamically linked sites of
20. I. Behm-Ansmant, J. Rehwinkel, T. Doerks, A. Stark, P. Bork, E. Izaur- mRNP remodeling. J. Cell Biol. 169, 871–884 (2005).
ralde, mRNA degradation by miRNAs and GW182 requires both 44. P. Anderson, N. Kedersha, RNA granules. J. Cell Biol. 172, 803–808 (2006).
CCR4:NOT deadenylase and DCP1:DCP2 decapping complexes. 45. M. K. Gross, G. F. Merrill, Thymidine kinase synthesis is repressed in
Genes Dev. 20, 1885–1898 (2006). nonreplicating muscle cells by a translational mechanism that does not
21. J. Rehwinkel, P. Natalin, A. Stark, J. Brennecke, S. M. Cohen, E. Izaur- affect the polysomal distribution of thymidine kinase mRNA. Proc. Natl.
ralde, Genome-wide analysis of mRNAs regulated by Drosha and Arg- Acad. Sci. U.S.A. 86, 4987–4991 (1989).
onaute proteins in Drosophila melanogaster. Mol. Cell. Biol. 26, 46. I. E. Clark, D. Wyckoff, E. R. Gavis, Synthesis of the posterior determi-
2965–2975 (2006). nant Nanos is spatially restricted by a novel cotranslational regulatory
22. R. C. Lee, R. L. Feinbaum, V. Ambros, The C. elegans heterochronic mechanism. Curr. Biol. 10, 1311–1314 (2000).
gene lin-4 encodes small RNAs with antisense complementarity to 47. L. P. Lim, N. C. Lau, P. Garrett-Engele, A. Grimson, J. M. Schelter, J.
lin-14. Cell 75, 843–854 (1993). Castle, D. P. Bartel, P. S. Linsley, J. M. Johnson, Microarray analysis
23. B. Wightman, I. Ha, G. Ruvkun, Posttranscriptional regulation of the shows that some microRNAs downregulate large numbers of target

Downloaded from stke.sciencemag.org on February 26, 2009


heterochronic gene lin-14 by lin-4 mediates temporal pattern formation mRNAs. Nature 433, 769–773 (2005).
in C. elegans. Cell 75, 855–862 (1993). 48. Q. Jing, S. Huang, S. Guth, T. Zarubin, A. Motoyama, J. Chen, F. Di
24. P. H. Olsen, V. Ambros, The lin-4 regulatory RNA controls developmen- Padova, S. C. Lin, H. Gram, J. Han, Involvement of microRNA in AU-rich
tal timing in Caenorhabditis elegans by blocking LIN-14 protein synthe- element-mediated mRNA instability. Cell 120, 623–634 (2005).
sis after the initiation of translation. Dev. Biol. 216, 671–680 (1999). 49. S. Bagga, J. Bracht, S. Hunter, K. Massirer, J. Holtz, R. Eachus, A. E.
25. K. Seggerson, L. Tang, E. G. Moss, Two genetic circuits repress the Pasquinelli, Regulation by let-7 and lin-4 miRNAs results in target
Caenorhabditis elegans heterochronic gene lin-28 after translation mRNA degradation. Cell 122, 553–563 (2005).
initiation. Dev. Biol. 243, 215–225 (2002). 50. L. Wu, J. Fan, J. G. Belasco, MicroRNAs direct rapid deadenylation of
26. J. E. Wilson, T. V. Pestova, C. U. Hellen, P. Sarnow, Initiation of protein mRNA. Proc. Natl. Acad. Sci. U.S.A. 103, 4034–4039 (2006).
synthesis from the A site of the ribosome. Cell 102, 511–520 (2000). 51. A. J. Giraldez, Y. Mishima, J. Rihel, R. J. Grocock, S. Van Dongen, K.
27. P. T. Nelson, A. G. Hatzigeorgiou, Z. Mourelatos, miRNP:mRNA association Inoue, A. J. Enright, A. F. Schier, Zebrafish miR-430 promotes dead-
in polyribosomes in a human neuronal cell line. RNA 10, 387–394 (2004). enylation and clearance of maternal mRNAs. Science 312, 75–79
28. J. Kim, A. Krichevsky, Y. Grad, G. D. Hayes, K. S. Kosik, G. M. Church, (2006).
G. Ruvkun, Identification of many microRNAs that copurify with polyri- 52. J. Ling, S. J. Morley, V. M. Pain, W. F. Marzluff, D. R. Gallie, The histone
bosomes in mammalian neurons. Proc. Natl. Acad. Sci. U.S.A. 101, 3′-terminal stem-loop-binding protein enhances translation through a
360–365 (2004). functional and physical interaction with eukaryotic initiation factor 4G
29. R. S. Pillai, S. N. Bhattacharyya, C. G. Artus, T. Zoller, N. Cougot, E. (eIF4G) and eIF3. Mol. Cell. Biol. 22, 7853–7867 (2002).
Basyuk, E. Bertrand, W. Filipowicz, Inhibition of translational initiation by 53. B. Gorgoni, S. Andrews, A. Schaller, D. Schumperli, N. K. Gray, B.
let-7 microRNA in human cells. Science 309, 1573–1576 (2005). Muller, The stem-loop binding protein stimulates histone translation at
30. S. N. Bhattacharyya, R. Habermacher, U. Martine, E. I. Closs, W. an early step in the initiation pathway. RNA 11, 1030–1042 (2005).
Filipowicz, Relief of microRNA-mediated translational repression in 54. J. Liu, M. A. Valencia-Sanchez, G. J. Hannon, R. Parker, MicroRNA-
human cells subjected to stress. Cell 125, 1111–1124 (2006). dependent localization of targeted mRNAs to mammalian P-bodies.
31. T. V. Pestova, C. U. Hellen, I. N. Shatsky, Canonical eukaryotic initiation Nat. Cell Biol. 7, 719–723 (2005).
factors determine initiation of translation by internal ribosomal entry. 55. G. L. Sen, H. M. Blau, Argonaute 2/RISC resides in sites of mammalian mR-
Mol. Cell. Biol. 16, 6859–6869 (1996). NA decay known as cytoplasmic bodies. Nat. Cell Biol. 7, 633–636 (2005).
32. T. V. Pestova, I. N. Shatsky, S. P. Fletcher, R. J. Jackson, C. U. Hellen, A 56. U. Sheth, R. Parker, Decapping and decay of messenger RNA occur in
prokaryotic-like mode of cytoplasmic eukaryotic ribosome binding to the cytoplasmic processing bodies. Science 300, 805–808 (2003).
initiation codon during internal translation initiation of hepatitis C and 57. D. Teixeira, U. Sheth, M. A. Valencia-Sanchez, M. Brengues, R. Parker,
classical swine fever virus RNAs. Genes Dev. 12, 67–83 (1998). Processing bodies require RNA for assembly and contain nontranslat-
33. D. T. Humphreys, B. J. Westman, D. I. Martin, T. Preiss, MicroRNAs ing mRNAs. RNA 11, 371–382 (2005).
control translation initiation by inhibiting eukaryotic initiation factor 58. M. Brengues, D. Teixeira, R. Parker, Movement of eukaryotic mRNAs
4E/cap and poly(A) tail function. Proc. Natl. Acad. Sci. U.S.A. 102, between polysomes and cytoplasmic processing bodies. Science 310,
16961–16966 (2005). 486–489 (2005).
34. V. G. Kolupaeva, T. V. Pestova, C. U. Hellen, I. N. Shatsky, Translation 59. T. Eystathioy, A. Jakymiw, E. K. Chan, B. Seraphin, N. Cougot, M. J. Frit-
eukaryotic initiation factor 4G recognizes a specific structural element zler, The GW182 protein colocalizes with mRNA degradation associat-
within the internal ribosome entry site of encephalomyocarditis virus ed proteins hDcp1 and hLSm4 in cytoplasmic GW bodies. RNA 9,
RNA. J. Biol. Chem. 273, 18599–18604 (1998). 1171–1173 (2003).
35. I. B. Lomakin, C. U. Hellen, T. V. Pestova, Physical association of 60. N. Cougot, S. Babajko, B. Seraphin, Cytoplasmic foci are sites of mRNA
eukaryotic initiation factor 4G (eIF4G) with eIF4A strongly enhances decay in human cells. J. Cell Biol. 165, 31–40 (2004).
binding of eIF4G to the internal ribosomal entry site of encephalomy- 61. M. A. Andrei, D. Ingelfinger, R. Heintzmann, T. Achsel, R. Rivera-Pomar,
ocarditis virus and is required for internal initiation of translation. Mol. R. Lührmann, A role for eIF4E and eIF4E-transporter in targeting
Cell. Biol. 20, 6019–6029 (2000). mRNPs to mammalian processing bodies. RNA 11, 717–727 (2005).
36. A. B. Sachs, P. Sarnow, M. W. Hentze, Starting at the beginning, middle 62. M. A. Ferraiuolo, S. Basak, J. Dostie, E. L. Murray, D. R. Schoenberg,
and end: Translation initiation in eukaryotes. Cell 89, 831–838 (1997). N. Sonenberg, A role for the eIF4E-binding protein in P-body formation
37. H. Imataka, A. Gradi, N. Sonenberg, A newly identified N-terminal and mRNA decay. J. Cell Biol. 170, 913–924 (2005).
amino acid sequence of human eIF4G binds poly(A)-binding protein 63. J. E. Wilhelm, M. Hilton, Q. Amos, W. Henzel, Cup is an eIF4E binding
and functions in poly(A)-dependent translation. EMBO J. 17, protein required for both the translational repression of oskar and the
7480–7489 (1998). recruitment of Barentsz. J. Cell Biol. 163, 1197–1204 (2003).
38. G. Bergamini, T. Preiss, M. W. Hentze, Picornavirus IRESes and the 64. A. Nakamura, K. Sato, K. Hanyu-Nakamura, Drosophila Cup is an
poly(A) tail jointly promote cap-independent translation in a mammalian eIF4E binding protein that associates with Bruno and regulates oskar
cell-free system. RNA 6, 1781–1790 (2000). mRNA translation in oogenesis. Dev. Cell 6, 69–78 (2004).
39. Y. M. Michel, A. M. Borman, S. Paulous, K. M. Kean, Eukaryotic initiation 65. M. R. Nelson, A. M. Leidal, C. A. Smibert, Drosophila Cup is an eIF4E-
factor 4G-poly(A) binding protein interaction is required for poly(A) tail- binding protein that functions in Smaug-mediated translational
mediated stimulation of picornavirus internal ribosome entry segment- repression. EMBO J. 23, 150–159 (2004).

www.stke.org/cgi/content/full/2007/367/re1 Page 12
REVIEW

66. A. Pause, G. Belsham, A.-C. Gingras, O. Donzé, T.-A. Lin, J. Lawrence Jr., 76. M. Chekulaeva, M. W. Hentze, A. Ephrussi, Bruno acts as a dual repres-
N. Sonenberg, Insulin-dependent stimulation of protein synthesis by phos- sor of oskar translation, promoting mRNA oligomerization and forma-
phorylation of a regulator of 5′-cap function. Nature 371, 762–767 (1994). tion of silencing particles. Cell 124, 521–533 (2006).
67. J. Liu, F. V. Rivas, J. Wohlschlegel, J. R. Yates III, R. Parker, G. J. Han- 77. J. Martinez, A. Patkaniowska, H. Urlaub, R. Luhrmann, T. Tuschl, Single-
non, A role for the P-body component GW182 in microRNA function. stranded antisense siRNAs guide target RNA cleavage in RNAi. Cell
Nat. Cell Biol. 7, 1261–1266 (2005). 110, 563–574 (2002).
68. A. Wilczynska, C. Aigueperse, M. Kress, F. Dautry, D. Weil, The transla- 78. J. Martinez, T. Tuschl, RISC is a 5′ phosphomonoester-producing RNA
tional regulator CPEB1 provides a link between Dcp1 bodies and stress endonuclease. Genes Dev. 18, 975–980 (2004).
granules. J. Cell Sci. 118, 981–992 (2005). 79. B. Wang, T. M. Love, M. E. Call, J. G. Doench, C. D. Novina, Recapitula-
69. J. Coller, R. Parker, General translational repression by activators of tion of short RNA-directed translational gene silencing in vitro. Mol. Cell
mRNA decapping. Cell 122, 875–886 (2005). 22, 553–560 (2006).
70. C. L. Jopling, M. Yi, A. M. Lancaster, S. M. Lemon, P. Sarnow, Modula- 80. S. P. Fletcher, I. K. Ali, A. Kaminski, P. Digard, R. J. Jackson, The influ-
tion of hepatitis C virus RNA abundance by a liver-specific microRNA. ence of viral coding sequences on pestivirus IRES activity reveals
Science 309, 1577–1581 (2005). fur ther parallels with translation initiation in prokaryotes. RNA 8,
71. C. Y. Chu, T. M. Rana, Translation repression in human cells by microR- 1558–1571 (2002).
NA-induced gene silencing requires RCK/p54. PLoS Biol. 4, e210 (2006). 81. P. A. Maroney, Y. Yu, J. Fisher, T. W. Nilsen, Evidence that microRNAs
72. J. Rehwinkel, I. Behm-Ansmant, D. Gatfield, E. Izaurralde, A crucial role are associated with translating messenger RNAs in human cells. Nat.
for GW182 and the DCP1:DCP2 decapping complex in miRNA-mediat- Struct. Mol. Biol. 13, 1102–1107 (2006).
ed gene silencing. RNA 11, 1640–1647 (2005). 82. S. Nottrott, M. J. Simard, J. D. Richter, Human let-7a miRNA blocks
73. N. Minshall, G. Thom, N. Standart, A conserved role of a DEAD box he- protein production on actively translating polyribosomes. Nat. Struct.
licase in mRNA masking. RNA 7, 1728–1742 (2001). Mol. Biol. 13, 1108–1114 (2006).
74. N. Minshall, N. Standart, The active form of Xp54 RNA helicase in 83. We thank V. Ambros, J. Belasco, E. Izaurralde, C. Jopling, E. Miska, C.
translational repression is an RNA-mediated oligomer. Nucleic Acids Novina, R. Parker, T. Preiss, and D. Scadden for helpful comments on
Res. 32, 1325–1334 (2004). the draft versions of this article.
75. A. Nakamura, R. Amikura, K. Hanyu, S. Kobayashi, Me31B silences
translation of oocyte-localising RNAs through the formation of cytoplas-

Downloaded from stke.sciencemag.org on February 26, 2009


mic RNP complex during Drosophila oogenesis. Development 128, Citation: R. J. Jackson, N. Standart, How do microRNAs regulate gene
3233–3242 (2001). expression? Sci. STKE 2007, re1 (2007).

www.stke.org/cgi/content/full/2007/367/re1 Page 13

You might also like